Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2995 2023-11-30 18:00:55 |
2 format correct Meta information modification 2995 2023-12-01 09:56:24 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhang, Z.; Zhan, F. Type 2 Cystatins in Human Immune and Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/52246 (accessed on 05 July 2024).
Zhang Z, Zhan F. Type 2 Cystatins in Human Immune and Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/52246. Accessed July 05, 2024.
Zhang, Zijun, Fenghuang Zhan. "Type 2 Cystatins in Human Immune and Cancer" Encyclopedia, https://encyclopedia.pub/entry/52246 (accessed July 05, 2024).
Zhang, Z., & Zhan, F. (2023, November 30). Type 2 Cystatins in Human Immune and Cancer. In Encyclopedia. https://encyclopedia.pub/entry/52246
Zhang, Zijun and Fenghuang Zhan. "Type 2 Cystatins in Human Immune and Cancer." Encyclopedia. Web. 30 November, 2023.
Type 2 Cystatins in Human Immune and Cancer
Edit

Type 2 cystatins are a group of small secreted protease inhibitors that regulate cysteine protease cathepsins and legumain. These enzymes regulate important cellular processes that are linked to the immune response and tumor progression, playing important roles in both autoimmune diseases and various types of cancers. Cysteine cathepsins are associated with the development of autoimmune diseases, tumor progression, and metastasis. Cystatins are categorized into three subfamilies: type 1, type 2, and type 3. The type 2 cystatin subfamily is the largest, containing 10 members, and consists entirely of small secreted proteins. Although type 2 cystatins have many shared biological roles, each member differs in structure, post-translational modifications (e.g., glycosylation), and expression in different cell types. These distinctions allow the type 2 cystatins to have unique biological functions and properties.

cystatin cathepsin protease inhibitor immunity inflammation tumor progression

1. Cathepsin, Legumain, and Cystatins as Immune Modulators

To introduce the immune-modulatory role of cystatins, it is important to also briefly cover the immunomodulatory role of their target proteins, which are cathepsins and legumain. Cathepsin C, S, L, K, and legumain are involved in autoimmune regulation and exhibit pro-inflammatory functions.
Cathepsin C regulates the maturation of neutrophil serine proteases (NSPs). The secretion of NSPs helps to degrade invading microorganisms and represents one of the major anti-bacterial mechanisms of neutrophils. NSPs also participate in inflammatory response regulation by proteolytically modifying or degrading extracellular inflammatory cytokines and chemokines and catalyzing the activation of inflammatory response-related receptors [1]. NSPs are initially generated as zymogens, which have a dipeptide structure at their N-terminus, keeping the protein in an inactive state. Cathepsin C cleaves this dipeptide to activate the NSPs [2]. In addition, cathepsin C regulates the activity of granzyme B, which is related to the immune response of CD8+ T cells and natural killer (NK) cells [3].
Cathepsin S is required for MHC class II antigen presentation since maturation of the MHC class II molecules depends on cathepsin S activity [4][5]. In antigen-presenting cells, the MHC-li chaperone complex is first generated. The li peptide in the MHC-li complex is a 10 kDa invariant peptide chain that masks the antigen binding site of MHC class II molecules, thereby inactivating the whole MHC class II molecule. The MHC-li complex is then delivered to the lysosome and the li peptide is cleaved by cathepsin S to activate the MHC class II molecule [6][7]. As a result, cathepsin S is mainly an immune modulator that enhances immune surveillance.
Cathepsin L is also involved in the maturation of MHC class II molecules in macrophages [8]. Additionally, cathepsin L is an activator of perforin, a key enzyme involved in pore formation by cytotoxic lymphocytes [3][9].
Cathepsin K, a collagenase that is selectively expressed in osteoclasts, is associated with autoimmune disease. It plays an important role in bone matrix remodeling and mediates the inflammatory stress on the bone surface by degrading collagen I, II, and elastin [4][10].
Legumain, as the target of CYSC, CYS6, and CYSF, also participates in immune regulation. Like cathepsin S and L, legumain promotes the maturation of MHC class II molecules and participates in the direct cleavage of antigens, thereby facilitating overall antigen presentation. Legumain can also activate Toll-like receptors, an upstream receptor of the Toll-like receptor signaling pathway, which are key players in the innate immune response [11].
Overall, cathepsins and legumain can trigger immune responses, and their dysregulation often correlates with the pathogenesis of autoimmune diseases, including rheumatic arthritis, systemic lupus, Sjogren’s syndrome, asthma, and psoriasis [4].
Since type 2 cystatins inhibit cathepsins and legumain, which in turn suppress inflammatory responses and immune cell activation, type 2 cystatins can be considered immune modulators. Indeed, most of the type 2 cystatins are immunosuppressive due, at least in part, to their ability to inhibit protease function. Type 2 cystatins play key roles in DC maturation and neutralizing cytotoxic lymphocyte cytotoxicity. Additionally, some of them can also act as ligands to activate anti-inflammatory pathways. The following subsections will talk about this in detail for each member of type 2 cystatins.

1.1. Cystatin SN (CYS1, CST1)

CYS1 contributes to allergic responses and the pathogenesis of allergic respiratory diseases. CYS1 and cystatin SA are highly expressed in the nasal epithelial surface of patients who have chronic rhinosinusitis (CRS) with nasal polyps. Using proteomics, researchers have discovered that the mucus of CRS patients contains high concentrations of CYS1 [12][13]. CYS1 is related to the Th2 immune response—an inflammation-related response that is highly associated with allergic responses and induced by IL-4. Currently, CYS1 serves as a biomarker for the activated Th2 immune response, which is mainly triggered by CD4+ T cells [14]. CST1 expression can be induced by IL-4 stimulation and is positively correlated with the upregulation of the Th2 immune response markers IL-33 and TSLP in eosinophilic CRS cases [15]. In addition, recombinant human CYS1 induces secretion of several Th2-related cytokines, including IL-5, IL-13, and IL-4, with an increase in Th2 cell infiltration [12]. For this reason, CST1 is a well-accepted biomarker for CRS diagnosis. Since CYS1 regulates the Th2 immune response, it is a good biomarker for asthma and allergic rhinitis. CST1 is upregulated in epithelial cells of the upper and lower airways in patients with asthma and allergic rhinitis [16][17]. The upregulation of CST1 is correlated with severe allergic respiratory disease [18][19]. Interestingly, allergic respiratory diseases are treated with corticosteroids which downregulate CST1 expression in the airway epithelial cells [16]. One study showed that CYS1 promotes the proliferation and migration of airway smooth muscle cells by activating the PI3K/AKT signaling pathway in an in vitro asthma model. This indicates that CYS1 participates in airway remodeling events and maybe a main contributor to asthma development [20].
Because CYS1 promotes the secretion of IL-4 in the immune cells, CYS1 may introduce an anti-inflammatory response in the microenvironment. Indeed, patients with CRS who have high CST1 expression in their nasal epithelial cells also have high levels of CCL18. CCL18 is an M2 macrophage polarizing cytokine that induces immunosuppression and anti-inflammatory responses [21][22]. In addition, CST1 plays an important role in introducing an anti-inflammatory response in acute liver failure (ALF) models [23]. CYS1 can directly bind to IFNGR1/2, acting as a competitive inhibitor of the pro-inflammatory cytokine IFN-γ. As a consequence, CYS1 inhibits the activation of the JAK/STAT1 pathway induced by IFN-γ and promotes the M2 polarization of macrophages in the liver [23]. In summary, CYS1 plays a role in the allergic Th2 immune response and anti-inflammatory processes.

1.2. Other SD-Type Cystatins (CST2, CST4, CST5, encoding CYS2, CYSS, CYSD)

In addition to CST1, clinical studies have shown that the other SD-type cystatins (CST2, CST4, and CST5) also participate in immune response regulation. To date, the mechanism behind their immune regulatory function is unknown. As a result, conclusions about their function in immune regulation cannot be drawn.
Although CST1 is the preferred clinical biomarker for allergic respiratory diseases, like asthma and CRS, CST2 can also be used as a clinical biomarker for allergic respiratory diseases [24][25][26]. CST4 is used as an anti-inflammatory marker since it is downregulated in rheumatic arthritis and Sjogren’s syndrome [27][28].
Unlike the other SD-type cystatins which are used as anti-inflammatory biomarkers, CST5 is used as an inflammatory biomarker [29][30][31]. An in vitro experiment on the human MRC-5 diploid lung cell transfection model showed that CYSD inhibits the replication of OC43 and 229e coronavirus strains at its physiological concentration, showing that CYSD has an anti-viral function [32].

1.3. Cystatin C (CYSC, CST3)

CYSC has been shown to have anti-inflammatory properties. Immature DCs express high levels of CST3, and this upregulation gradually disappears during DC maturation [33]. The expression of CYSC protein in immature DCs prevents the protease activity of cathepsin S from activating the MHC class II molecules. Therefore, immature DCs are less able to complete antigen presentation and a following immune response [34]. A high level of CYSC positively correlates with the severity of several autoimmune diseases, including sepsis and rheumatoid arthritis [35][36]. A high level of CYSC also correlates with more severe HIV infections, which can be suppressed by antiviral treatment [37].
On the other hand, CYSC also shows pro-inflammatory activities. In hematopoietic cells, CYSC is expressed at higher levels in macrophages and DCs than in T cells [38][39]. In macrophages, CYSC modulates the immune response by enhancing macrophage responses to IFN-γ. This consequently upregulates the activation of the NF-κB pathway and promotes immune-related cytokine NO and TNF-α secretion [40]. CYSC expression can inhibit the secretion of the anti-inflammatory cytokine IL-10 and it is an antagonist of TGF-β [41][42]. At the same time, IL-10 and TGF-β can also modulate the expression of CYSC via the upregulation of its transcription factor IRF-8, forming a negative feedback loop [40][41].

1.4. Cystatin E/M (CYS6, CST6)

The immunomodulatory role of CYS6 has not been reported in the current literature. However, some predictions can be made based on the current mechanistic studies of CYS6. CYS6 can inhibit the activation of NF-κB signaling by both canonical and non-canonical pathways [43][44]. CYS6 can also be internalized by macrophages where it highly suppresses osteoclast cathepsin K [44]. Given this evidence, CYS6 has the potential for development as a therapeutic agent to alleviate rheumatic arthritis [4].

1.5. Cystatin F (CYSF, CST7)

CYSF is an important immune modulator that acts as a natural immunosuppressant in the human body. High levels of CYSF in NK cells, T cells, and DC cells have been used to counteract the activated immune response mediated by cathepsins. A high level of CYSF reduces the cytotoxicity of CD8+ T cells and NK cells. The inhibition of cathepsin C, L, and H in NK cells and CD8+ T cells suppresses the expression of the downstream cytotoxic-related protein perforin and granzymes [45][46]. In contrast, CYSF co-localizes with cathepsin S in immature DCs and weakens as the DCs mature [47]. The inhibition of cathepsin S by CYSF in immature DCs likely suppresses antigen presentation induced by the MHC class II molecules in DCs. Studies suggest that CYSF is associated with inflammatory responses in the central nervous system induced by microglia, a type of cell that is closely related to macrophages in the neural system [48][49].
In conclusion, besides CYSD and CYSC’s non-protease inhibitory function, the type 2 cystatins are all anti-inflammatory factors and are potential immunosuppressants.

2. The Role of Cathepsins, Legumain, and Type 2 Cystatins in Cancer Development

Type 2 cystatins are cysteine cathepsin inhibitors and this is also the reason why they have immunosuppressive effects. Theoretically, type 2 cystatins should have pro-tumor effects on cancer models as they are immunosuppressants. However, the situation is complex because several cathepsins also play roles in tumorigenesis-enhancing cellular events. The activity of cathepsin B, a ubiquitous cathepsin, is highly related to tumor progression and various devastating immunosuppressive events. These events include reduction in CD8+ T cell persistence, infiltration of immunosuppressive tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs) [4][50]. In addition, cathepsin B, L, S, and K participate in the cleavage and degradation of the extracellular matrix (ECM). As a result, the activation of these cathepsins facilitates the migration, invasion, and metastasis of tumor cells [51]. Cathepsins also have a protective effect on tumor cells and are involved in the chemoresistance of cancer.
Legumain is largely expressed on TAMs and cancer cells from the breast, prostate, and liver. The overexpression of legumain is highly correlated with tumor migration, invasion, poor prognosis, and inferior survival. Legumain can activate MMP-2&9, PI3K/AKT, and integrin signaling pathways to promote epithelial-mesenchymal transition (EMT) and the TGF-β signaling pathway. Additionally, it also cleaves and inactivates the tumor suppressor protein p53 [52].
Since type 2 cystatins inhibit both protease-induced inflammatory responses and pro-tumor activity, they exhibit a complex dual effect on tumorigenesis. In the following subsections, the role of each type 2 cystatin in cancer development is discussed.

2.1. Cystatin SN (CYS1)

CST1 is a pro-tumor gene in multiple cancer models, including esophageal, breast, colon, gastric, liver, and lung cancer [53][54][55][56][57][58]. In lung cancer, CST1 is hypomethylated [59], resulting in upregulation of CST1. This upregulation is often correlated with poor prognosis, metastasis, and recurrence [54][55][57][58]. Compared to CYSC, CYS1 is a weaker inhibitor but it displays a higher affinity for cathepsin B, which is known to be involved in tumor progression. Therefore, overexpression of CYS1 can partially neutralize the inhibitory effect of CYSC on cathepsin B, inducing stronger tumor invasiveness mediated by enhanced cathepsin B activity [60]. In addition to its ability to neutralize CYSC, CYS1 also upregulates AKT phosphorylation and activates the PI3K/AKT pathway. This subsequently downregulates E-cadherin and facilitates EMT, promoting tumor metastasis [58]. CYS1 directly interacts with the ferroptosis mediator GPX4 by interfering with the ubiquitination of GPX4 and stabilizing it, thus inhibiting tumor cell ferroptosis. This consequently promotes tumor progression and metastasis in gastric cancer models [53].

2.2. Other SD-Type Cystatins (CST2, CST4, CST5)

Since SD-type cystatins share a highly similar amino acid sequence and protein structure, it is not surprising that CYS2 and CYSS exhibit pro-tumor functions. However, studies on these cystatins are limited, which makes it difficult to conclude whether they have the same pro-tumor mechanism as CYS1. Upregulation of CST2 correlates with tumor metastasis of prostate and gastric cancer [61][62]. CYS2 may help regulate the TGF-β signaling pathway and promote EMT like CYS1, but the mechanism needs further exploration to be certain [61]. CST4 is upregulated in esophageal, colon, and gastric cancer and is associated with poor prognosis [63][64][65]. The overexpression of CYSS in gastric tumor cells upregulates a protein called ELFN2, which directly inhibits the expression of E-cadherin and promotes EMT, like CYS1 and CYS2 [64]. However, there are very few studies on ELFN2 and its connection with tumor invasiveness. Thus, future studies are needed to determine how CYSS promotes tumor growth.
In contrast, based on biomarker studies of colon, gastric, and prostate cancer, CST5 is considered an antitumor gene [66][67][68]. CST5 expression is induced by vitamin D, which is known to have antitumor activity and is downregulated in human colon cancer cells [69][70][71]. CYSD inhibits the Wnt/β-catenin signaling pathway and oncogenic c-MYC expression, which consequently extends the cell cycle and reduces the proliferation, migration, and invasiveness of colorectal tumor cells [69]. Importantly, some studies have suggested that CST5 is a downstream target of p53 [72]. The p53 protein enhances CST5 expression by interacting with the CST5 promoter region and simultaneously downregulating EMT-promoting transcription factor SNAIL, which in turn downregulates CST5 expression [72].

2.3. Cystatin C (CYSC)

CYSC has a dual effect on cancer development. It was observed to have an antitumor function on several cancer types, including pancreatic cancer, breast cancer, and leukemia [73][74][75][76]. Currently, there are at least two mechanisms that can explain the antitumor function of CYSC. First, the antitumor effect of CYSC is highly related to its strong inhibitory effect on cathepsin B. Compared with other type 2 cystatins, CYSC is the strongest inhibitor of cathepsin B [77]. Because CYSC inhibits cathepsin B, CYSC acts as a suppressor of cell migration, thereby inhibiting tumor invasion and metastasis [78]. CYSC is also an inhibitor of the TGF-β pathway. CYSC directly interacts with TGF-β receptor 2 and competes with the binding of the TGF-β ligand. By inhibiting the TGF- β signaling pathway, CYSC inhibits multiple metastatic events in tumor cells, such as loss of cell contacts, downregulation of cell polarization, and increased cell migration [79].
In contrast, there are also several studies stating that a high CYSC level correlates with a worse prognosis. High levels of CYSC were found in tumor tissue of ovary, colon, and esophageal cancer [80][81][82]. Cathepsin B has been found to promote apoptosis and this could be linked to the observed phenomenon that CYSC suppresses apoptosis of tumor cells [83][84]. However, the mechanism by which CYSC promotes tumor progression is unknown. CYSC can be used as a biomarker for cancer prognosis. However, recent studies suggest that renal function, which is frequently altered in cancer, heavily influences the levels of serum CYSC and intercellular CYSC [85][86]. This could negatively impact the use of CYSC as an effective biomarker for cancer prognosis.

2.4. Cystatin E/M (CYS6)

CYS6 has been found to have dual effects, exhibiting pro-tumor properties in some cancers and anti-tumor properties in others. CST6 is an antitumor gene that is hypermethylated in breast, prostate, brain, and cervical cancer [43][87][88][89]. The antitumor effect of CST6 has been mostly studied in breast cancer models and its downregulation correlates with higher levels of migration and invasiveness in both in vivo and in vitro models of breast cancer [87]. The epigenetic silencing of CST6 is strongly correlated to breast cancer bone metastasis [87]. CYS6 also inhibits the pro-oncogenic function of legumain [90][91]. In addition, CYS6 inhibits cathepsin B, which is frequently upregulated in breast cancer [92]. Cathepsin B cleaves SPHK1, an enzyme that inhibits osteoclast differentiation by inhibiting p38 activation induced by RANKL [92]. Uncontrolled osteoclast differentiation disrupts bone homeostasis, causes bone disease, and promotes cancer bone metastasis [93]. Furthermore, CYS6 inhibits cathepsin K, an enzyme that is predominantly expressed in osteoclasts and participates in bone matrix remodeling [44]. Therefore, CYS6 suppresses bone metastasis of tumor cells by inhibiting osteoclastogenesis. CYS6 also inhibits NF-κB signaling in both canonical and non-canonical pathways [43][44].
In contrast, CST6 has also been found to have pro-tumor functions in pancreatic, liver, gastric, and triple-negative breast cancer [94][95][96][97]. Additionally, a small fraction of patients with multiple myeloma express high levels of CST6 [44]. A recent paper has identified CST6 as a factor involved in the dysregulation of necroptosis in gastric tumor cells, resulting in a poor prognosis for patients with gastric cancer [97].

2.5. Cystatin F (CYSF)

Although there are extensive reports on the immunoregulatory function of CYSF, the role of CYSF in cancer is unclear. Given the immunosuppressive function of CYSF, uncontrolled upregulation of CST7 likely enhances tumor progression. Indeed, high levels of CST7 are associated with poor prognosis and lower survival rates in patients with liver, oral, and brain cancer [46][98][99]. The current literature has shown that the pro-tumor effects of CYSF are highly correlated to its immunosuppressive function. Upregulation of CST7 causes a decrease in the cytotoxicity of NK cells to tumor cells [98]. In contrast, multiple studies suggest that CST7 downregulation is associated with higher invasiveness of tumors, more metastatic events, and tumor progression in prostate cancer, lung cancer, pancreatic cancer, and lymphoma [100][101][102][103]. Since CST7 downregulation is correlated with metastasis and tumor progression, this indicates that CST7, like other type 2 cystatin family members, regulates the cathepsin B-L metastatic axis. However, further studies are needed to draw definitive conclusions on the role of CST7 in cancer progression.

References

  1. Pham, C.T. Neutrophil serine proteases: Specific regulators of inflammation. Nat. Rev. Immunol. 2006, 6, 541–550.
  2. Shen, X.B.; Chen, X.; Zhang, Z.Y.; Wu, F.F.; Liu, X.H. Cathepsin C inhibitors as anti-inflammatory drug discovery: Challenges and opportunities. Eur. J. Med. Chem. 2021, 225, 113818.
  3. Kos, J.; Nanut, M.P.; Prunk, M.; Sabotic, J.; Dautovic, E.; Jewett, A. Cystatin F as a regulator of immune cell cytotoxicity. Cancer Immunol. Immunother. 2018, 67, 1931–1938.
  4. Senjor, E.; Kos, J.; Nanut, M.P. Cysteine Cathepsins as Therapeutic Targets in Immune Regulation and Immune Disorders. Biomedicines 2023, 11, 476.
  5. Shi, G.P.; Villadangos, J.A.; Dranoff, G.; Small, C.; Gu, L.; Haley, K.J.; Riese, R.; Ploegh, H.L.; Chapman, H.A. Cathepsin S required for normal MHC class II peptide loading and germinal center development. Immunity 1999, 10, 197–206.
  6. Riese, R.J.; Wolf, P.R.; Bromme, D.; Natkin, L.R.; Villadangos, J.A.; Ploegh, H.L.; Chapman, H.A. Essential role for cathepsin S in MHC class II-associated invariant chain processing and peptide loading. Immunity 1996, 4, 357–366.
  7. Driessen, C.; Bryant, R.A.; Lennon-Dumenil, A.M.; Villadangos, J.A.; Bryant, P.W.; Shi, G.P.; Chapman, H.A.; Ploegh, H.L. Cathepsin S controls the trafficking and maturation of MHC class II molecules in dendritic cells. J. Cell. Biol. 1999, 147, 775–790.
  8. Nakagawa, T.Y.; Brissette, W.H.; Lira, P.D.; Griffiths, R.J.; Petrushova, N.; Stock, J.; McNeish, J.D.; Eastman, S.E.; Howard, E.D.; Clarke, S.R.; et al. Impaired invariant chain degradation and antigen presentation and diminished collagen-induced arthritis in cathepsin S null mice. Immunity 1999, 10, 207–217.
  9. House, I.G.; House, C.M.; Brennan, A.J.; Gilan, O.; Dawson, M.A.; Whisstock, J.C.; Law, R.H.; Trapani, J.A.; Voskoboinik, I. Regulation of perforin activation and pre-synaptic toxicity through C-terminal glycosylation. EMBO Rep. 2017, 18, 1775–1785.
  10. Garnero, P.; Borel, O.; Byrjalsen, I.; Ferreras, M.; Drake, F.H.; McQueney, M.S.; Foged, N.T.; Delmas, P.D.; Delaisse, J.M. The collagenolytic activity of cathepsin K is unique among mammalian proteinases. J. Biol. Chem. 1998, 273, 32347–32352.
  11. Solberg, R.; Lunde, N.N.; Forbord, K.M.; Okla, M.; Kassem, M.; Jafari, A. The Mammalian Cysteine Protease Legumain in Health and Disease. Int. J. Mol. Sci. 2022, 23, 15983.
  12. Nocera, A.L.; Mueller, S.K.; Workman, A.D.; Wu, D.; McDonnell, K.; Sadow, P.M.; Amiji, M.M.; Bleier, B.S. Cystatin SN is a potent upstream initiator of epithelial-derived type 2 inflammation in chronic rhinosinusitis. J. Allergy Clin. Immunol. 2022, 150, 872–881.
  13. Yan, B.; Lou, H.; Wang, Y.; Li, Y.; Meng, Y.; Qi, S.; Wang, M.; Xiao, L.; Wang, C.; Zhang, L. Epithelium-derived cystatin SN enhances eosinophil activation and infiltration through IL-5 in patients with chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2019, 144, 455–469.
  14. Higham, A.; Dungwa, J.; Pham, T.H.; McCrae, C.; Singh, D. Increased mast cell activation in eosinophilic chronic obstructive pulmonary disease. Clin. Transl. Immunol. 2022, 11, e1417.
  15. Kato, Y.; Takabayashi, T.; Sakashita, M.; Imoto, Y.; Tokunaga, T.; Ninomiya, T.; Morikawa, T.; Yoshida, K.; Noguchi, E.; Fujieda, S. Expression and Functional Analysis of CST1 in Intractable Nasal Polyps. Am. J. Respir. Cell. Mol. Biol. 2018, 59, 448–457.
  16. Wang, M.; Gong, L.; Luo, Y.; He, S.; Zhang, X.; Xie, X.; Li, X.; Feng, X. Transcriptomic analysis of asthma and allergic rhinitis reveals CST1 as a biomarker of unified airways. Front. Immunol. 2023, 14, 1048195.
  17. Jakiela, B.; Soja, J.; Sladek, K.; Przybyszowski, M.; Plutecka, H.; Gielicz, A.; Licholai, S.; Aab, A.; Rebane, A.; Bochenek, G. Bronchial epithelial cell transcriptome shows endotype heterogeneity of asthma in patients with NSAID-exacerbated respiratory disease. J. Allergy Clin. Immunol. 2023, 151, 953–965.
  18. Kack, U.; Einarsdottir, E.; van Hage, M.; Asarnoj, A.; James, A.; Nopp, A.; Krjutskov, K.; Katayama, S.; Kere, J.; Lilja, G.; et al. Nasal upregulation of CST1 in dog-sensitised children with severe allergic airway disease. ERJ Open Res. 2021, 7.
  19. Wu, D.; Yan, B.; Wang, Y.; Wang, C.; Zhang, L. Prognostic and pharmacologic value of cystatin SN for chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2021, 148, 450–460.
  20. Liu, H.B.; Bai, J.; Wang, W.X. CST1 promotes the proliferation and migration of PDGF-BB-treated airway smooth muscle cells via the PI3K/AKT signaling pathway. Kaohsiung J. Med. Sci. 2023, 39, 145–153.
  21. Nakayama, T.; Lee, I.T.; Le, W.; Tsunemi, Y.; Borchard, N.A.; Zarabanda, D.; Dholakia, S.S.; Gall, P.A.; Yang, A.; Kim, D.; et al. Inflammatory molecular endotypes of nasal polyps derived from White and Japanese populations. J. Allergy Clin. Immunol. 2022, 149, 1296–1308.
  22. Schraufstatter, I.U.; Zhao, M.; Khaldoyanidi, S.K.; Discipio, R.G. The chemokine CCL18 causes maturation of cultured monocytes to macrophages in the M2 spectrum. Immunology 2012, 135, 287–298.
  23. Xu, Y.; Wang, J.; Ren, H.; Dai, H.; Zhou, Y.; Ren, X.; Wang, Y.; Feng, S.; Deng, X.; Wu, J.; et al. Human endoderm stem cells reverse inflammation-related acute liver failure through cystatin SN-mediated inhibition of interferon signaling. Cell Res. 2023, 33, 147–164.
  24. Miyake, M.M.; Workman, A.D.; Nocera, A.L.; Wu, D.; Mueller, S.K.; Finn, K.; Amiji, M.M.; Bleier, B.S. Discriminant analysis followed by unsupervised cluster analysis including exosomal cystatins predict presence of chronic rhinosinusitis, phenotype, and disease severity. Int. Forum Allergy Rhinol. 2019, 9, 1069–1076.
  25. Mueller, S.K.; Wendler, O.; Nocera, A.; Grundtner, P.; Schlegel, P.; Agaimy, A.; Iro, H.; Bleier, B.S. Escalation in mucus cystatin 2, pappalysin-A, and periostin levels over time predict need for recurrent surgery in chronic rhinosinusitis with nasal polyps. Int. Forum Allergy Rhinol. 2019, 9, 1212–1219.
  26. Workman, A.D.; Nocera, A.L.; Mueller, S.K.; Otu, H.H.; Libermann, T.A.; Bleier, B.S. Translating transcription: Proteomics in chronic rhinosinusitis with nasal polyps reveals significant discordance with messenger RNA expression. Int. Forum Allergy Rhinol. 2019, 9, 776–786.
  27. Boto de Los Bueis, A.; de la Fuente, M.; Montejano-Milner, R.; Del Hierro Zarzuelo, A.; Vecino, E.; Acera, A. A Pilot Study of a Panel of Ocular Inflammation Biomarkers in Patients with Primary Sjogren’s Syndrome. Curr. Issues Mol. Biol. 2023, 45, 2881–2894.
  28. Soria, J.; Acera, A.; Merayo, L.J.; Duran, J.A.; Gonzalez, N.; Rodriguez, S.; Bistolas, N.; Schumacher, S.; Bier, F.F.; Peter, H.; et al. Tear proteome analysis in ocular surface diseases using label-free LC-MS/MS and multiplexed-microarray biomarker validation. Sci. Rep. 2017, 7, 17478.
  29. Elam, M.B.; Majumdar, G.; Mozhui, K.; Gerling, I.C.; Vera, S.R.; Fish-Trotter, H.; Williams, R.W.; Childress, R.D.; Raghow, R. Patients experiencing statin-induced myalgia exhibit a unique program of skeletal muscle gene expression following statin re-challenge. PLoS ONE 2017, 12, e0181308.
  30. Cowell, W.; Colicino, E.; Lee, A.G.; Bosquet Enlow, M.; Flom, J.D.; Berin, C.; Wright, R.O.; Wright, R.J. Data-driven discovery of mid-pregnancy immune markers associated with maternal lifetime stress: Results from an urban pre-birth cohort. Stress 2020, 23, 349–358.
  31. Panezai, J.; Ali, A.; Ghaffar, A.; Benchimol, D.; Altamash, M.; Klinge, B.; Engstrom, P.E.; Larsson, A. Upregulation of circulating inflammatory biomarkers under the influence of periodontal disease in rheumatoid arthritis patients. Cytokine 2020, 131, 155117.
  32. Collins, A.R.; Grubb, A. Cystatin D, a natural salivary cysteine protease inhibitor, inhibits coronavirus replication at its physiologic concentration. Oral Microbiol. Immunol. 1998, 13, 59–61.
  33. Zavasnik-Bergant, T.; Repnik, U.; Schweiger, A.; Romih, R.; Jeras, M.; Turk, V.; Kos, J. Differentiation- and maturation-dependent content, localization, and secretion of cystatin C in human dendritic cells. J. Leukoc. Biol. 2005, 78, 122–134.
  34. Pierre, P.; Mellman, I. Developmental regulation of invariant chain proteolysis controls MHC class II trafficking in mouse dendritic cells. Cell 1998, 93, 1135–1145.
  35. Biasizzo, M.; Trstenjak-Prebanda, M.; Dolinar, K.; Pirkmajer, S.; Zavrsnik, J.; Turk, B.; Kopitar-Jerala, N. Cystatin C Deficiency Increases LPS-Induced Sepsis and NLRP3 Inflammasome Activation in Mice. Cells 2021, 10, 2071.
  36. Hansen, T.; Petrow, P.K.; Gaumann, A.; Keyszer, G.; Brauer, R.; Kriegsmann, J. Synovial giant cells in rheumatoid arthritis: Expression of cystatin C, but not of cathepsin B. Exp. Toxicol. Pathol. 2000, 52, 312–316.
  37. Longenecker, C.T.; Hileman, C.O.; Funderburg, N.T.; McComsey, G.A. Rosuvastatin preserves renal function and lowers cystatin C in HIV-infected subjects on antiretroviral therapy: The SATURN-HIV trial. Clin. Infect. Dis. 2014, 59, 1148–1156.
  38. Xu, Y.; Lindemann, P.; Vega-Ramos, J.; Zhang, J.G.; Villadangos, J.A. Developmental regulation of synthesis and dimerization of the amyloidogenic protease inhibitor cystatin C in the hematopoietic system. J. Biol. Chem. 2014, 289, 9730–9740.
  39. El-Sukkari, D.; Wilson, N.S.; Hakansson, K.; Steptoe, R.J.; Grubb, A.; Shortman, K.; Villadangos, J.A. The protease inhibitor cystatin C is differentially expressed among dendritic cell populations, but does not control antigen presentation. J. Immunol. 2003, 171, 5003–5011.
  40. Frendeus, K.H.; Wallin, H.; Janciauskiene, S.; Abrahamson, M. Macrophage responses to interferon-gamma are dependent on cystatin C levels. Int. J. Biochem. Cell. Biol. 2009, 41, 2262–2269.
  41. Xu, Y.; Schnorrer, P.; Proietto, A.; Kowalski, G.; Febbraio, M.A.; Acha-Orbea, H.; Dickins, R.A.; Villadangos, J.A. IL-10 controls cystatin C synthesis and blood concentration in response to inflammation through regulation of IFN regulatory factor 8 expression. J. Immunol. 2011, 186, 3666–3673.
  42. Sokol, J.P.; Schiemann, W.P. Cystatin C antagonizes transforming growth factor beta signaling in normal and cancer cells. Mol. Cancer Res. 2004, 2, 183–195.
  43. Soh, H.; Venkatesan, N.; Veena, M.S.; Ravichandran, S.; Zinabadi, A.; Basak, S.K.; Parvatiyar, K.; Srivastava, M.; Liang, L.J.; Gjertson, D.W.; et al. Cystatin E/M Suppresses Tumor Cell Growth through Cytoplasmic Retention of NF-kappaB. Mol. Cell Biol. 2016, 36, 1776–1792.
  44. Gai, D.; Chen, J.R.; Stewart, J.P.; Nookaew, I.; Habelhah, H.; Ashby, C.; Sun, F.; Cheng, Y.; Li, C.; Xu, H.; et al. CST6 suppresses osteolytic bone disease in multiple myeloma by blocking osteoclast differentiation. J. Clin. Investig. 2022, 132, e159527.
  45. Prunk, M.; Nanut, M.P.; Sabotic, J.; Svajger, U.; Kos, J. Increased cystatin F levels correlate with decreased cytotoxicity of cytotoxic T cells. Radiol. Oncol. 2019, 53, 57–68.
  46. Magister, S.; Tseng, H.C.; Bui, V.T.; Kos, J.; Jewett, A. Regulation of split anergy in natural killer cells by inhibition of cathepsins C and H and cystatin F. Oncotarget 2015, 6, 22310–22327.
  47. Magister, S.; Obermajer, N.; Mirkovic, B.; Svajger, U.; Renko, M.; Softic, A.; Romih, R.; Colbert, J.D.; Watts, C.; Kos, J. Regulation of cathepsins S and L by cystatin F during maturation of dendritic cells. Eur. J. Cell. Biol. 2012, 91, 391–401.
  48. Jin, C.; Shao, Y.; Zhang, X.; Xiang, J.; Zhang, R.; Sun, Z.; Mei, S.; Zhou, J.; Zhang, J.; Shi, L. A Unique Type of Highly-Activated Microglia Evoking Brain Inflammation via Mif/Cd74 Signaling Axis in Aged Mice. Aging Dis. 2021, 12, 2125–2139.
  49. Mangale, V.; Syage, A.R.; Ekiz, H.A.; Skinner, D.D.; Cheng, Y.; Stone, C.L.; Brown, R.M.; O’Connell, R.M.; Green, K.N.; Lane, T.E. Microglia influence host defense, disease, and repair following murine coronavirus infection of the central nervous system. Glia 2020, 68, 2345–2360.
  50. Ma, K.; Chen, X.; Liu, W.; Chen, S.; Yang, C.; Yang, J. CTSB is a negative prognostic biomarker and therapeutic target associated with immune cells infiltration and immunosuppression in gliomas. Sci. Rep. 2022, 12, 4295.
  51. Fonovic, M.; Turk, B. Cysteine cathepsins and extracellular matrix degradation. Biochim. Biophys. Acta. 2014, 1840, 2560–2570.
  52. Mai, C.W.; Chung, F.F.; Leong, C.O. Targeting Legumain As a Novel Therapeutic Strategy in Cancers. Curr. Drug Targets 2017, 18, 1259–1268.
  53. Li, D.; Wang, Y.; Dong, C.; Chen, T.; Dong, A.; Ren, J.; Li, W.; Shu, G.; Yang, J.; Shen, W.; et al. CST1 inhibits ferroptosis and promotes gastric cancer metastasis by regulating GPX4 protein stability via OTUB1. Oncogene 2023, 42, 83–98.
  54. Dai, D.N.; Li, Y.; Chen, B.; Du, Y.; Li, S.B.; Lu, S.X.; Zhao, Z.P.; Zhou, A.J.; Xue, N.; Xia, T.L.; et al. Elevated expression of CST1 promotes breast cancer progression and predicts a poor prognosis. J. Mol. Med. 2017, 95, 873–886.
  55. Cao, X.; Li, Y.; Luo, R.Z.; Zhang, L.; Zhang, S.L.; Zeng, J.; Han, Y.J.; Wen, Z.S. Expression of Cystatin SN significantly correlates with recurrence, metastasis, and survival duration in surgically resected non-small cell lung cancer patients. Sci. Rep. 2015, 5, 8230.
  56. Zhang, L.; Yu, S.; Yin, X.; Tu, M.; Cai, L.; Zhang, Y.; Yu, L.; Zhang, S.; Pan, X.; Huang, Y. MiR-942-5p inhibits tumor migration and invasion through targeting CST1 in esophageal squamous cell carcinoma. PLoS ONE 2023, 18, e0277006.
  57. Li, T.; Xiong, Q.; Zou, Z.; Lei, X.; Jiang, Q.; Liu, D. Prognostic significance of cystatin SN associated nomograms in patients with colorectal cancer. Oncotarget 2017, 8, 115153–115163.
  58. Cui, Y.; Sun, D.; Song, R.; Zhang, S.; Liu, X.; Wang, Y.; Meng, F.; Lan, Y.; Han, J.; Pan, S.; et al. Upregulation of cystatin SN promotes hepatocellular carcinoma progression and predicts a poor prognosis. J. Cell Physiol. 2019, 234, 22623–22634.
  59. Mullapudi, N.; Ye, B.; Suzuki, M.; Fazzari, M.; Han, W.; Shi, M.K.; Marquardt, G.; Lin, J.; Wang, T.; Keller, S.; et al. Genome Wide Methylome Alterations in Lung Cancer. PLoS ONE 2015, 10, e0143826.
  60. Kim, J.T.; Lee, S.J.; Kang, M.A.; Park, J.E.; Kim, B.Y.; Yoon, D.Y.; Yang, Y.; Lee, C.H.; Yeom, Y.I.; Choe, Y.K.; et al. Cystatin SN neutralizes the inhibitory effect of cystatin C on cathepsin B activity. Cell Death Dis. 2013, 4, e974.
  61. Zhang, W.P.; Wang, Y.; Tan, D.; Xing, C.G. Cystatin 2 leads to a worse prognosis in patients with gastric cancer. J. Biol. Regul. Homeost. Agents 2020, 34, 2059–2067.
  62. Song, F.; Zhang, Y.; Pan, Z.; Hu, X.; Yi, Y.; Zheng, X.; Wei, H.; Huang, P. Identification of novel key genes associated with the metastasis of prostate cancer based on bioinformatics prediction and validation. Cancer Cell. Int. 2021, 21, 559.
  63. Shi, D.; Zhou, Z.; Zhang, S. miRNA-6715-5p Inhibits Cellular Proliferation and Invasion in Colorectal Cancer by Directly Targeting CST4. J. Oncol. 2021, 2021, 7615712.
  64. Zhang, Y.Q.; Zhang, J.J.; Song, H.J.; Li, D.W. Overexpression of CST4 promotes gastric cancer aggressiveness by activating the ELFN2 signaling pathway. Am. J. Cancer Res. 2017, 7, 2290–2304.
  65. Yang, G.; Zhang, Y.; Lin, H.; Liu, J.; Huang, S.; Zhong, W.; Peng, C.; Du, L. CircRNA circ_0023984 promotes the progression of esophageal squamous cell carcinoma via regulating miR-134-5p/cystatin-s axis. Bioengineered 2022, 13, 10578–10593.
  66. Wu, W.; Yong, W.W.; Chung, M.C. A simple biomarker scoring matrix for early gastric cancer detection. Proteomics 2016, 16, 2921–2930.
  67. Berger, M.D.; Stintzing, S.; Heinemann, V.; Cao, S.; Yang, D.; Sunakawa, Y.; Matsusaka, S.; Ning, Y.; Okazaki, S.; Miyamoto, Y.; et al. A Polymorphism within the Vitamin D Transporter Gene Predicts Outcome in Metastatic Colorectal Cancer Patients Treated with FOLFIRI/Bevacizumab or FOLFIRI/Cetuximab. Clin. Cancer Res. 2018, 24, 784–793.
  68. Munetsuna, E.; Kawanami, R.; Nishikawa, M.; Ikeda, S.; Nakabayashi, S.; Yasuda, K.; Ohta, M.; Kamakura, M.; Ikushiro, S.; Sakaki, T. Anti-proliferative activity of 25-hydroxyvitamin D3 in human prostate cells. Mol. Cell Endocrinol. 2014, 382, 960–970.
  69. Alvarez-Diaz, S.; Valle, N.; Garcia, J.M.; Pena, C.; Freije, J.M.; Quesada, V.; Astudillo, A.; Bonilla, F.; Lopez-Otin, C.; Munoz, A. Cystatin D is a candidate tumor suppressor gene induced by vitamin D in human colon cancer cells. J. Clin. Investig. 2009, 119, 2343–2358.
  70. Deeb, K.K.; Trump, D.L.; Johnson, C.S. Vitamin D signalling pathways in cancer: Potential for anticancer therapeutics. Nat. Rev. Cancer 2007, 7, 684–700.
  71. Carlberg, C.; Munoz, A. An update on vitamin D signaling and cancer. Semin. Cancer Biol. 2022, 79, 217–230.
  72. Hunten, S.; Hermeking, H. p53 directly activates cystatin D/CST5 to mediate mesenchymal-epithelial transition: A possible link to tumor suppression by vitamin D3. Oncotarget 2015, 6, 15842–15856.
  73. Wang, B.; Sun, J.; Kitamoto, S.; Yang, M.; Grubb, A.; Chapman, H.A.; Kalluri, R.; Shi, G.P. Cathepsin S controls angiogenesis and tumor growth via matrix-derived angiogenic factors. J. Biol. Chem. 2006, 281, 6020–6029.
  74. Balaji, K.N.; Schaschke, N.; Machleidt, W.; Catalfamo, M.; Henkart, P.A. Surface cathepsin B protects cytotoxic lymphocytes from self-destruction after degranulation. J. Exp. Med. 2002, 196, 493–503.
  75. Indacochea, A.; Guerrero, S.; Urena, M.; Araujo, F.; Coll, O.; ME, L.L.; Gebauer, F. Cold-inducible RNA binding protein promotes breast cancer cell malignancy by regulating Cystatin C levels. RNA 2021, 27, 190–201.
  76. Mori, J.; Tanikawa, C.; Funauchi, Y.; Lo, P.H.; Nakamura, Y.; Matsuda, K. Cystatin C as a p53-inducible apoptotic mediator that regulates cathepsin L activity. Cancer Sci. 2016, 107, 298–306.
  77. Abrahamson, M.; Barrett, A.J.; Salvesen, G.; Grubb, A. Isolation of six cysteine proteinase inhibitors from human urine. Their physicochemical and enzyme kinetic properties and concentrations in biological fluids. J. Biol. Chem. 1986, 261, 11282–11289.
  78. Nakabayashi, H.; Hara, M.; Shimuzu, K. Clinicopathologic significance of cystatin C expression in gliomas. Hum. Pathol. 2005, 36, 1008–1015.
  79. Sokol, J.P.; Neil, J.R.; Schiemann, B.J.; Schiemann, W.P. The use of cystatin C to inhibit epithelial-mesenchymal transition and morphological transformation stimulated by transforming growth factor-beta. Breast Cancer Res. 2005, 7, R844–R853.
  80. Saleh, Y.; Sebzda, T.; Warwas, M.; Kopec, W.; Ziolkowska, J.; Siewinski, M. Expression of cystatin C in clinical human colorectal cancer tissues. J. Exp. Ther. Oncol. 2005, 5, 49–53.
  81. Periyasamy, A.; Gopisetty, G.; Subramanium, M.J.; Velusamy, S.; Rajkumar, T. Identification and validation of differential plasma proteins levels in epithelial ovarian cancer. J. Proteom. 2020, 226, 103893.
  82. Yan, Y.; Fan, Q.; Wang, L.; Zhou, Y.; Li, J.; Zhou, K. LncRNA Snhg1, a non-degradable sponge for miR-338, promotes expression of proto-oncogene CST3 in primary esophageal cancer cells. Oncotarget 2017, 8, 35750–35760.
  83. Vancompernolle, K.; Van Herreweghe, F.; Pynaert, G.; Van de Craen, M.; De Vos, K.; Totty, N.; Sterling, A.; Fiers, W.; Vandenabeele, P.; Grooten, J. Atractyloside-induced release of cathepsin B, a protease with caspase-processing activity. FEBS Lett. 1998, 438, 150–158.
  84. Decock, J.; Obermajer, N.; Vozelj, S.; Hendrickx, W.; Paridaens, R.; Kos, J. Cathepsin B, cathepsin H, cathepsin X and cystatin C in sera of patients with early-stage and inflammatory breast cancer. Int. J. Biol. Markers 2008, 23, 161–168.
  85. Lameire, N.; Vanholder, R.; Van Biesen, W.; Benoit, D. Acute kidney injury in critically ill cancer patients: An update. Crit. Care 2016, 20, 209.
  86. Rosner, M.H.; Perazella, M.A. Acute Kidney Injury in Patients with Cancer. N. Engl. J. Med. 2017, 376, 1770–1781.
  87. Jin, L.; Zhang, Y.; Li, H.; Yao, L.; Fu, D.; Yao, X.; Xu, L.X.; Hu, X.; Hu, G. Differential secretome analysis reveals CST6 as a suppressor of breast cancer bone metastasis. Cell. Res. 2012, 22, 1356–1373.
  88. Pulukuri, S.M.; Gorantla, B.; Knost, J.A.; Rao, J.S. Frequent loss of cystatin E/M expression implicated in the progression of prostate cancer. Oncogene 2009, 28, 2829–2838.
  89. Qiu, J.; Ai, L.; Ramachandran, C.; Yao, B.; Gopalakrishnan, S.; Fields, C.R.; Delmas, A.L.; Dyer, L.M.; Melnick, S.J.; Yachnis, A.T.; et al. Invasion suppressor cystatin E/M (CST6): High-level cell type-specific expression in normal brain and epigenetic silencing in gliomas. Lab Investig. 2008, 88, 910–925.
  90. D’Costa, Z.C.; Higgins, C.; Ong, C.W.; Irwin, G.W.; Boyle, D.; McArt, D.G.; McCloskey, K.; Buckley, N.E.; Crawford, N.T.; Thiagarajan, L.; et al. TBX2 represses CST6 resulting in uncontrolled legumain activity to sustain breast cancer proliferation: A novel cancer-selective target pathway with therapeutic opportunities. Oncotarget 2014, 5, 1609–1620.
  91. Wallin, H.; Apelqvist, J.; Andersson, F.; Ekstrom, U.; Abrahamson, M. Low-level internalization of cystatin E/M affects legumain activity and migration of melanoma cells. J. Biol. Chem. 2017, 292, 14413–14424.
  92. Li, X.; Liang, Y.; Lian, C.; Peng, F.; Xiao, Y.; He, Y.; Ma, C.; Wang, Y.; Zhang, P.; Deng, Y.; et al. CST6 protein and peptides inhibit breast cancer bone metastasis by suppressing CTSB activity and osteoclastogenesis. Theranostics 2021, 11, 9821–9832.
  93. Fornetti, J.; Welm, A.L.; Stewart, S.A. Understanding the Bone in Cancer Metastasis. J. Bone Min. Res. 2018, 33, 2099–2113.
  94. Li, Q.; Zheng, Z.C.; Ni, C.J.; Jin, W.X.; Jin, Y.X.; Chen, Y.; Zhang, X.H.; Chen, E.D.; Cai, Y.F. Correlation of Cystatin E/M with Clinicopathological Features and Prognosis in Triple-Negative Breast Cancer. Ann. Clin. Lab. Sci. 2018, 48, 40–44.
  95. Zhou, X.; Wang, X.; Huang, K.; Liao, X.; Yang, C.; Yu, T.; Liu, J.; Han, C.; Zhu, G.; Su, H.; et al. Investigation of the clinical significance and prospective molecular mechanisms of cystatin genes in patients with hepatitis B virus-related hepatocellular carcinoma. Oncol. Rep. 2019, 42, 189–201.
  96. Hosokawa, M.; Kashiwaya, K.; Eguchi, H.; Ohigashi, H.; Ishikawa, O.; Furihata, M.; Shinomura, Y.; Imai, K.; Nakamura, Y.; Nakagawa, H. Over-expression of cysteine proteinase inhibitor cystatin 6 promotes pancreatic cancer growth. Cancer Sci. 2008, 99, 1626–1632.
  97. Khan, M.; Lin, J.; Wang, B.; Chen, C.; Huang, Z.; Tian, Y.; Yuan, Y.; Bu, J. A novel necroptosis-related gene index for predicting prognosis and a cold tumor immune microenvironment in stomach adenocarcinoma. Front. Immunol. 2022, 13, 968165.
  98. Senjor, E.; Perisic Nanut, M.; Breznik, B.; Mitrovic, A.; Mlakar, J.; Rotter, A.; Porcnik, A.; Lah Turnsek, T.; Kos, J. Cystatin F acts as a mediator of immune suppression in glioblastoma. Cell. Oncol. 2021, 44, 1051–1063.
  99. Utsunomiya, T.; Hara, Y.; Kataoka, A.; Morita, M.; Arakawa, H.; Mori, M.; Nishimura, S. Cystatin-like metastasis-associated protein mRNA expression in human colorectal cancer is associated with both liver metastasis and patient survival. Clin. Cancer Res. 2002, 8, 2591–2594.
  100. Chahal, M.S.; Ku, H.T.; Zhang, Z.; Legaspi, C.M.; Luo, A.; Hopkins, M.M.; Meier, K.E. Differential Expression of Ccn4 and Other Genes Between Metastatic and Non-metastatic EL4 Mouse Lymphoma Cells. Cancer Genom. Proteom. 2016, 13, 437–442.
  101. Yang, C.; Yu, T.; Liu, Z.; Ye, X.; Liao, X.; Wang, X.; Han, C.; Zhu, G.; Qin, W.; Peng, T. Cystatin F as a key family 2 cystatin subunit and prognostic biomarker for early-stage pancreatic ductal adenocarcinoma. Oncol. Rep. 2019, 42, 79–90.
  102. Baniwal, S.K.; Khalid, O.; Gabet, Y.; Shah, R.R.; Purcell, D.J.; Mav, D.; Kohn-Gabet, A.E.; Shi, Y.; Coetzee, G.A.; Frenkel, B. Runx2 transcriptome of prostate cancer cells: Insights into invasiveness and bone metastasis. Mol. Cancer 2010, 9, 258.
  103. Qi, Q.; Chen, C.; Liu, C.; Zhang, B.; Ma, Y.; Zhang, H.; Huang, W.; Wang, C. Linc8087 predicts favorable prognosis and inhibits cell migration and invasion in NSCLC. Pathol. Res. Pract. 2021, 225, 153569.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 158
Revisions: 2 times (View History)
Update Date: 01 Dec 2023
1000/1000
Video Production Service